Sei sulla pagina 1di 9

Cytometry Part B (Clinical Cytometry) 90B:31–39 (2016)

Original Article
Consensus Guidelines on Plasma Cell Myeloma
Minimal Residual Disease Analysis and Reporting
Maria Arroz,1* Neil Came,2 Pei Lin,3 Weina Chen,4 Constance Yuan,5 Anand Lagoo,6
Mariela Monreal,7 Ruth de Tute,8 Jo-Anne Vergilio,9 Andy C. Rawstron,10
and Bruno Paiva11
1
Department of Clinical Pathology, Cytometry Laboratory, CHLO, Hospital S. Francisco Xavier, Lisbon, Portugal
2
Pathology Department, Peter MacCallum Cancer Centre, Melbourne, Australia
3
Department of Hematopathology, MD Anderson Cancer Center, Houston, Texas, USA
4
Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
5
Laboratory of Pathology, NCI, NIH, Bethesda, Maryland, USA
6
Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
7
Interpflow Corporation, Miami, Florida, USA
8
HMDS, Department of Haematology, St. James’s Institute of Oncology, Leeds, United Kingdom
9
University Of Michigan Medical Center Hematology Oncology Laboratory, Ann Arbor, Michigan, USA
10
HMDS, Department of Haematology, St. James’s Institute of Oncology, Leeds, United Kingdom
11
Clinica Universidad de Navarra, Centro de Investigaci
on Medica Aplicada, University of Navarra, Pamplona, Spain

Background: Major heterogeneity between laboratories in flow cytometry (FC) minimal residual disease
(MRD) testing in multiple myeloma (MM) must be overcome. Cytometry societies such as the International Clin-
ical Cytometry Society and the European Society for Clinical Cell Analysis recognize a strong need to establish
minimally acceptable requirements and recommendations to perform such complex testing.
Methods: A group of 11 flow cytometrists currently performing FC testing in MM using different instru-
mentation, panel designs ( 6-color) and analysis software compared the procedures between their
respective laboratories and reviewed the literature to propose a consensus guideline on flow-MRD analy-
sis and reporting in MM.
Results/Conclusion: Consensus guidelines support i) the use of minimum of five initial gating parameters
(CD38, CD138, CD45, forward, and sideward light scatter) within the same aliquot for accurate identification of
the total plasma cell compartment; ii) the analysis of potentially aberrant phenotypic markers and to report the
antigen expression pattern on neoplastic plasma cells as being reduced, normal or increased, when compared
to a normal reference plasma cell immunophenotype (obtained using the same instrument and parameters);
and iii) the percentage of total bone marrow plasma cells plus the percentages of both normal and neoplastic
plasma cells within the total bone marrow plasma cell compartment, and over total bone marrow cells. Consen-
sus guidelines on minimal current and future MRD analyses should target a lower limit of detection of
0.001%, and ideally a limit of quantification of 0.001%, which requires at least 3 3 106 and 5 3 106 bone
marrow cells to be measured, respectively. VC 2015 International Clinical Cytometry Society

Key terms: plasma cell myeloma; minimal residual disease; flow cytometry

How to cite this article: Arroz M, Came N, Lin P, Chen W, Yuan C, Lagoo A, Monreal M, de Tute R, Vergilio
J.-A, Rawstron A C. and Paiva B. Consensus Guidelines on Plasma Cell Myeloma Minimal Residual Disease Analy-
sis and Reporting. Cytometry Part B 2016; 90B: 31–39.

*Correspondence to: Maria Arroz, Department of Clinical Pathology, Received 9 December 2014; Revised 15 January 2015; Accepted
Cytometry Laboratory, CHLO, Hospital S. Francisco Xavier, Estrada do 16 January 2015
Forte do Alto do Duque, 1495-005 Lisbon, Portugal. E-mail: marroz@ Published online 2 June 2015 in Wiley Online Library
chlo.min-saude.pt (wileyonlinelibrary.com).
DOI: 10.1002/cyto.b.21228

C 2015 International Clinical Cytometry Society


V
32 ARROZ ET AL.

FIG. 1. Initial gating strategy for identifying the BM PC compartment. A relevant parameter is plotted against time as a measure of the quality and con-
tinuity of data acquired for the whole sample (A). Light scatter characteristics are used to exclude doublets (B and C) and debris (D). All possible plasma
cells are captured by generous gating on CD381 and CD1381 bright events (E) and CD381 bright events plotted against CD45 (F).

The principles outlined in the Practice Guidelines that tions such as sequential or Boolean gating, hierarchical
were recently published by the International Council for clustering, or principle component analysis (PCA) are
Standardization in Haematology (ICSH) and International now commonly available, and will further help on iden-
Clinical Cytometry Society (ICCS) (1) guided an interna- tifying rare plasma cell (PC) events, in a reproducible
tional working group of experts in the field, under the way, within pluricellular bone marrow (BM) samples.
auspices of the ICCS and the European Society for Clini-
cal Cell Analysis (ESCCA) toward consensus on: analysis FLOW CYTOMETRY DATA ANALYSIS AND PLASMA CELL
and gating strategy, data interpretation, reporting con- GATING STRATEGY
ventions, and a minimum set of recommendations that In adopting a “fit for purpose” gating strategy for iden-
can be applied in the optimal assessment of minimal tifying potentially rare PC events it is important to use a
residual disease (MRD) in multiple myeloma (MM) using gating strategy which minimizes subjectivity and maxi-
multidimensional flow cytometry (FC). mizes reproducibility (1). Inicial gates to identify PCs
An increasingly higher number of flow cytometry lab- and discriminate them from all other BM cells are drawn
oratories use digital instruments that can accommodate generously enough to include as many PCs events as
eight or more colors. These provide superior event possible, allowing for variants that express lower CD38,
acquisition and analysis of a greater number of cells uti- CD45 and/or CD138 to be included, while excluding
lizing more parameters per tube than was previously fea- contaminating lymphocytes, doublets and other cellular
sible with four color instruments. Six well-chosen colors debris (Fig. 1).
in a single tube represent the minimum number recom- Overall despite different instruments and variable
mended for low-sensitive flow-MRD analysis in MM. Simi- flow-MRD panel designs (6, 8, or 10-color) between lab-
larly, all currently available analysis software packages oratories, the adoption of the European Myeloma Net
enable the drawing of regions (“gates”) allowing the par- (EMN) approach using a minimum of five gating parame-
titioning of data into cell subsets in a relatively user- ters (CD38, CD138, CD45, FSC, and SSC properties)
friendly, stepwise, accurate, and reproducible way. More within the same tube for the identification of the total
sophisticated “downstream” algorithms that require PC compartment is recommended (2). This maximizes
rapid execution of complex mathematical transforma- the inclusion of normal and neoplastic PCs whilst, at

Cytometry Part B: Clinical Cytometry


GUIDELINES ON MYELOMA MRD ANALYSIS AND REPORTING 33

FIG. 2. Expression of routinely used antigens on bone marrow aspirates. A: Erythroblasts and leukocytes by SSC vs. CD45, (B) Normal B-cells/hem-
atogones by SSC vs. CD19; (C) CD27 expression on memory T and B-cells by SSC vs. CD27; (D) CD81 expression on B-cells/hematogones by SSC
vs. CD81; (E) NK cells by SSC vs. CD56; (F) mast cells, myeloid, and early erythroid progenitors by SSC vs. CD117.

the same time, allowing removal of contaminating non- exclusion since both normal and neoplastic PCs can
PC events. (2). show variable CD45 expression.

Initial Gating Strategy


Further Characterization of PCs
The recommended gating steps are as follows (Fig. 1):
Albeit heterogeneous, the normal BM PC immunophe-
i. Use of a key parameter (e.g., CD38) versus time to notype is highly conserved in normal bone marrow sam-
assess the quality and continuity of data acquired, ples, as well as between individual patient samples at
which provides, for example, the opportunity to gate different disease stages, except when antibody-based
out air bubbles and clogs, thus eliminating periods of therapies have been used (e.g.: anti-CD38; 3). In this
time of unstable flow (A). regard, based on merged data files from different sam-
ii. Use of light scatter to exclude doublets, (FSC-Height ples, followed by principle component analysis of the
vs. FSC-Area) (B), (SSC-Height vs. SSC-Area) (C), and immunophenotype of normal BM PCs (as assessed by up
debris, (SSC-Area vs. FSC-Area) (D) while exercising to 12 different markers combined in two eight-color
caution not to exclude, from initial gating, hyperdi- tubes (CD45, CD138, CD38, CD19, CD56, or CD28,
ploid or tetraploid PCs which exhibit aberrantly high B2M, or CD27, CyIgj or CD117, CyIgk or CD81), the
FSC-Area and SSC-Area, but, e.g. low, FSC-Height. Euroflow Consortium demonstrated that BM PCs from
iii. Generous gating is then performed to encompass all healthy donors, patients with non-PC related diseases, or
potential CD381 and CD1381 PCs (E), which is fur- regenerating BM following chemotherapy in patients
ther refined by incorporating all CD381 bright with non-PC related diseases, had fully overlapping and
events plotted against CD45 (F). CD45 is helpful for highly similar immunophenotypes (3). In addition, such
gating but should not be used as a marker for cell normal PC immunophenotypes did not overlap with

Cytometry Part B: Clinical Cytometry


34 ARROZ ET AL.

FIG. 3. Normal and neoplastic PCs coexisting within the BM PC compartment. A and B demonstrate the combined analysis gate strategy to identify
all PCs. According to multiple abnormalities in antigen expression, neoplastic PCs (black) and normal PCs (grey) can be identified in subsequent dot
plots for CD56 vs. CD38 (C), CD45 vs. CD19 (D), CD81 vs. CD117 (E), and CD38 vs. CD27 (F).

that of >97% of neoplastic PCs from a large series of subsets showing myeloma-like aberrant phenotypes (e.g.
MM patients studied at diagnosis. CD19-negative or CD56-positive PCs), can be a valuable
That notwithstanding, it should be noted that appropri- second step to confirm the normal vs. aberrant nature of
ate multivariate analysis with  8 markers is required to such PCs in a minor subset of cases.
reach such an applicability because it is now well Even though antigenic shift upon therapy cannot be
recognized that minor subsets of normal BM PCs can precluded (7), major antigenic shifts with respect to the
express some markers with intensity levels previously consensus markers were typically absent in large
considered to be aberrant (4,5,6). In this regard, Liu et al. cohorts in the Medical Research Council Myeloma IX
(5) and Tembhare et al. (6) have proposed some helpful Study (8). However, it is important to note that the base-
insights that can assist in the discrimination between nor- line tumor clone often shows also phenotypic heteroge-
mal versus neoplastic PCs, as described below. neity, and that all different phenotypic subclones should
Normal PCs show heterogeneous expression of CD19 be followed throughout therapy (9). In fact, it has been
and CD45, they are usually negative for CD20 and CD117, recently shown that the distribution of such subclones
and they invariably show homogeneously bright expres- may fluctuate from diagnostic into MRD samples, but
sion of CD81 (6). In turn, CD28 and CD56 expression is this should be interpreted as clonal selection rather than
present in a small subset (between 5–20%) of normal PCs. antigenic shift upon therapy (9).
Accordingly, normal PCs typically show heterogeneous or
bimodal expression for some of the most commonly used Defining Neoplastic PCs
markers (e.g. for CD19, CD56, and CD45) while neoplas- No single parameter reliably distinguishes neoplastic
tic PCs from MM patients usually have more homogene- PCs from normal PCs; however, even a limited (six
ous antigen expression. Neoplastic PCs will often show color) combination of antibodies in a single tube is able
additional aberrancies, such as CD117 expression, weaker to achieve this in a high proportion of cases provided a
CD38 expression, and increased FSC and/or SSC features. sufficient number of cellular events are evaluated in the
Noteworthy, such aberrancies will be co-expressed within flow cytometer (8). The comparison between the
the same cell population, whereas immunophenotypic patient’s sample vs. a normal PC immunophenotype ref-
variations in normal PCs tend to be heterogeneous and/or erence obtained using the same instrument and parame-
distributed amongst different well-defined subsets. Pro- ters may be highly valuable. Similarly, internal positive
vided a sufficient number of PCs are acquired, analysis of (e.g., B-cells for CD19 and CD81, NK cells for CD56,
cytoplasmic light chain expression within specific PCs myeloid progenitors and mast cells for CD117) and

Cytometry Part B: Clinical Cytometry


GUIDELINES ON MYELOMA MRD ANALYSIS AND REPORTING 35

FIG. 4. A: Example of a principle component analysis (PCA) guided approach as the starting point for the separation of a homogeneous PC popula-
tion (black) from the remaining bone marrow cells (grey) traced onto CD38 vs. CD138 (B) and CD38 vs. CD45 (C) dot plots, using the same sample
as depicted in Figure 1. D: Another example of PCA, in this case separating neoplastic PCs (black) from normal PCs (grey) restricted to the total pre-
viously gated bone marrow plasma cell compartment, as demonstrated in figure 3 A and B, traced onto a bivariate plot (E) analyzing CD38 vs.
CD27.

negative control (e.g., erythroblasts for CD45) cell popu- antibodies, under identical sample preparation proce-
lations represent a highly reliable strategy to define nor- dures and conditions (12). In order to accommodate the
mal vs. aberrant phenotypic profiles. This should also be degree of intra and inter-patient phenotypic heterogeneity
performed using the patient’s sample for each relevant of normal PCs, the larger the library of cases, the better. A
antigen as illustrated in Figure 2. minimum number of twenty reference cases (for which
The 2006 Bethesda International Consensus Recommen- similar cell numbers to those of the questioned samples
dations advise on the reporting of antigen expression as have been evaluated), is recommended. The discrimina-
being reduced, normal, or increased in intensity compared tion between normal PCs and neoplastic PCs on pheno-
with that expressed on appropriate reference cell popula- typic grounds has to be multiparametric, i.e. the identifi-
tions (10). Reporting in this manner provides a clearer pic- cation of a neoplastic PC population should never be
ture of the immunophenotypic deviation from normal PC based on the isolated deviation of a single marker.
immunophenotype and minimizes the impact of variability As described above, analysis of the CyIgj/CyIgk
in reagent and instrument performance between laborato- plasma cell ratio within specific PC subsets defined on
ries. Reporting should include the percentage of positive the basis of other surface markers (e.g.: CD19- CD45-
cells for each marker within the specific neoplastic PC CD561) may be informative to confirm the clonal vs.
compartment. polyclonal nature of such PC subsets in selected cases.
The availability of patient-specific diagnostic immuno-
phenotypes may also represent a valuable aid while per- Summary of the Most Frequent Normal and Neoplastic PC
forming flow-MRD monitoring. The use of polychromatic Immunophenotypes, and of Less Frequent Phenotypes
(8-color) flow cytometry strategies facilitates this Observed on Minor Subsets of Normal Plasma Cells
approach (11). The most common immunophenotype of normal PCs
Perhaps even more useful, particularly in the setting of could be described as CD381bright, CD1381bright,
multicenter trials, is the availability of a reference library CD191, CD451, CD202, CD271, CD282, CD562,
of normal and neoplastic BM PCs that have been immuno- CD811, CD1172, CD2002/1, with a polyclonal CyIgj/
phenotyped using the exact same panel of monoclonal CyIgk ratio. Minor subsets of normal PCs (typically

Cytometry Part B: Clinical Cytometry


36 ARROZ ET AL.

representing <30% of total normal PCs) exhibit less classification of normal vs. malignant PCs, and be an
“typical” antigen expression patterns such as: CD192, ideal adjunct towards myeloma MRD assessment and
CD452/low, CD201, CD27low, CD281, CD561 and standardization by flow cytometry.
CD2001 bright. (2,13,14).
In contrast, neoplastic PCs show aberrant phenotypes REPORTING CONVENTIONS
consisting of multiple possible combinations of the follow- Reporting the overall immunophenotypic signature of
ing aberrant antigenic profiles: CD38 low (dimmer than neoplastic PCs
normal PCs), CD192, CD452, CD201, CD272/low,
CD281, CD561/bright, CD812/low, CD1171, CD200 Reporting an antigen expression pattern on neoplastic
bright, in association with monoclonal CyIgj or CyIgk. PCs on an individual marker basis as being reduced, nor-
Figure 3 illustrates an example where coexisting normal mal or increased, compared to a normal (reference) PC
and neoplastic PCs within the same bone marrow, display immunophenotype (obtained using the same antibody-
distinct phenotypic features. reagent combinations, instruments and sample prepara-
tion protocols and conditions), including the percentage
The role of new/more advanced computational tools to of positive cells for each marker, is recommended to
facilitate data analysis for flow-MRD in MM establish the immunophenotypic signature that will aid
Traditional visualization of flow cytometric data in in follow-up MRD detection.
bivariate (2D) dot plots results in an increasingly large
number of scattergrams and a “geometric increase in the Reporting a sample as MRD positive, MRD negative, or
informational content” as the number of antibodies used unsuitable for MRD assessment
increases (10). This is a particular challenge for users The guidelines of the EMN in 2008 proposed that an
becoming familiar with 8 color flow cytometry and MRD negative BM is defined as no detectable abnormal
requires the judicious use and graphical representation of PCs at a level of sensitivity of at least 0.01%. However
all potentially relevant 2D combinations of antigen this is on the proviso that the BM aspirate sample is
expression patterns. There is a risk that by relying on vis- deemed suitable using the criteria defined earlier, by vis-
ual interpretation of two-dimensional plots alone the ualizing all BM cells simultaneously (Fig. 2). An MRD-
operator may not fully appreciate all relevant antigen negative bone marrow, in the appropriate clinical con-
expression patterns within a particular cell population, text, has also been termed as immunophenotypic
nor gain full insight into the variation of normal antigen response (IR) or flow-Complete Response (F-CR) (18).
expression profiles. In recent years, a variety of new However, the threshold used to distinguish MRD-positive
computational tools have been developed to identify spe- from MRD-negative may vary from sample to sample,
cific cell populations in multidimensional flow cytometry and is subject to change as technology and treatments
data sets (15,16). One such solution is the use of princi- improve; therefore, it is recommended to report on the
pal component analysis (PCA) of a unified data file with a level of MRD and the assay limit of detection in addition
potentially unlimited number of dimensions (17). A PCA- to classifying patients as MRD-negative vs. MRD-positive.
based approach is implemented in Infinicyt software in At present, several clinical studies have validated the
the automatic population separator (APS) view, where prognostic value of myeloma flow-MRD monitoring. In
the first (x axis) and second (y axis) principal compo- some studies, the presence of MRD was evaluated quanti-
nents are used to produce a bidimensional representation tatively as percent of neoplastic PCs from all BM
of either cell clusters or patient profiles; each principal nucleated cells (e.g., CD451 leucocytes plus CD45- cellu-
component is represented by a linear combination of all lar BM events such as nucleated red bood cells) (19,20).
parameters included in the FCS file(s) under analysis, as Other studies have used the number of total leukocytes,
examplified in Figure 4. The power of this interpretation defined as PCs plus non-PCs CD45 positive events as the
method is that it facilitates the development of a normal denominator (8). Reporting PC levels as a percentage of
reference library of chosen cases, automated separation nucleated cells is likely to offer a better comparison with
of populations within a sample taking into consideration molecular techniques than reporting as a percentage of
all parameters, and prospective detection and tracking of leucocytes because DNA is typically isolated from all
any aberrant population that deviates from the normal nucleated cells. However, different lysing approaches (e.g.
cells. This can be particularly useful in cases with differ- ammonium chloride vs. fixative-based lysing approaches)
ent subclones that can be difficult to identify when only can affect the number of total nucleated cells because
bi-dimensional dot plots are used (9). ammonium chloride will also lyse some nucleated ery-
While not all users have practical experience with throid cells, while using total leucocytes as the denomina-
these new multidimensional data analysis tools and tor is not affected by different lysing agents. The impact
approaches, or they are not presently resourced to of different reporting or lysing approaches is likely to
adopt it, automated multivariate (e.g. PCA-guided) data have a negligible effect on the level of MRD detected but
analysis approaches for fast identification and characteri- the report must clearly state which method is used.
zation of homogeneous cell populations, coupled with Although multicentre data has shown that while using
reference (normal and tumor) databases, in the context 0.01%/1024 as the limit of detection (LOD) for MRD
of full technical standardization, may provide automated monitoring by flow is of clear prognostic value, it does

Cytometry Part B: Clinical Cytometry


GUIDELINES ON MYELOMA MRD ANALYSIS AND REPORTING 37

Table 1 could be considered to be below the limit of detection is


Estimated LOD and LLOQ According to the Total Number of 19 events, but due to the potential counting errors (95%
Events Acquired confidence interval for a count of 19 events would be of
Total number of 11 to 30 events) this implies that the LOD can be estimated
cells analyzed LOD (%) LLOQ (%) as (30/total number of cells analyzed) 3 100%. Similarly, it
100,000 0.03 0.05 is also widely accepted that more than 50 events is a stand-
200,000 0.015 0.025 ard threshold for reproducible enumeration of a cell popu-
500,000 0.006 0.01 lation by experienced flow cytometer operators (28,29);
1,000,000 0.003 0.005 consequently, the limit of quantification can be estimated
2,000,000 0.0015 0.0025
3,000,000 0.001 0.0017 as (50/total number of cells analyzed) 3 100%. Thus, the
5,000,000 0.0006 0.001 LOD and the LLOQ will be both typically dependent on
the total number of cells analyzed; the expected limit of
LOD 5 (30/Total cells) 3100%; LOQ 5 (50/Total cells) 3100%. detection and lower limit of quantification for a range of
cells analyzed are shown in Table 1.
not yet translate into a plateau (8,19,21). In MM as in Reporting both the number of neoplastic PCs identi-
other diseases, it is almost certain that as treatment regi- fied in a sample and the total number of (normal 1 neo-
mens continue to improve, the threshold of 0.01%/1024 plastic) events contained in it, is mandatory when
is likely to become less relevant. The consensus is that expressing the level of sensitivity of the flow-MRD assay
current and future MRD analyses should target a lower performed in an individual sample. Thus, a complete
LOD preferable of < 0.001%/1025 (18,22), which report would include the percentage of neoplastic PCs
requires acquisition of at least 3 3 106 bone marrow from the whole sample cellularity, either the LOD or
cells; ideally a limit of quantification of 0.001% should LLOQ or both, depending on the MRD levels, and ideally
be targeted in the new MM flow-MRD approaches which also the total number of cells evaluated for that sample,
requires that a minimum of 5 3 106 BM cells are after excluding doublets and debris. For example, if neo-
acquired. If it is required that a categorical assessment plastic PCs are not detected, then the minimal data
(MRD-positive or MRD-negative) is provided, it is impor- required includes the LOD of the assay, including ideally
tant to state the threshold used and ideally reference the also the total number of cells evaluated, e.g.: neoplastic
threshold appropriately, e.g. MRD-negative at the 0.01% PCs < 0.0004% (neoplastic PCs < 20 events; total cells
level [EMN guidelines (2)]. analyzed 5 5,000,000). In turn, if neoplastic PCs are
detected above the LLOQ, then the minimal data
Reporting MRD levels and the limit of detection (LOD) and required is the percentage of neoplastic PCs and the
the lower limit of quantification (LLOQ) LLOQ, ideally including also the number of events (neo-
As discussed above, the use of statements such as MRD- plastic PCs and total cells) measured, e.g.: neoplastic
positive or MRD-negative can be poorly informative if PCs 5 0.05% of the cells (LLOQ 0.0025%; neoplastic
there is not a universally agreed threshold defined for PCs 5 1,000 events; total cells analyzed 5 2,000,000).
(uniform) response criteria. Although many laboratories Finally if neoplastic PCs are detected at a level between
are currently performing more sensitive flow-MRD assays that of the LOD and the LLOQ, then a percentage MRD
(i. e.:0.001%/1025), a threshold of 0.01%/1024 is currently cannot be reported; instead the lower and upper limits
accepted as being the minimum clinically relevant and are reported, e.g.: neoplastic PCs detected below the
routinely achievable threshold. In order to ensure that quantitative range (0.0004 2 0.001%; neoplastic
MRD results are informative, even if the accepted thresh- PCs 5 35 events, total cells analyzed 5 5,000,000). The
old for MRD detection will likely change in the future proportion of neoplastic PCs within the total plasma
(e.g. down to 0.001%/1025), it is critical that the MRD cell compartment should always be reported for two
report includes sufficient information to determine the reasons: first, the information might be of prognostic
detection and quantification limits of the individual assay, relevance (30); secondly, because the balance between
used for any individual sample. Such limits are based on phenotypically aberrant and normal PCs is not likely to
four major parameters: (i) the total number of events ana- be affected significantly by hemodilution, therefore pro-
lyzed, (ii) the number of neoplastic PC events detected viding a measure about the quality of the sample.
within them, (iii) the smallest number of events required
to reproducibly detect a population of neoplastic PCs SUMMARY LIST OF POTENTIAL PITFALLS OCCURRING
and, (iv) the smallest number of events required to repro- DURING MRD DATA ANALYSIS
ducibly quantify a population of neoplastic PCs.
The College of American Pathologists requires that each
laboratory confirm the lower LOD and LLOQ for each test  Gating errors associated with inclusion of non-plasma
(23–25). There are numerous studies demonstrating that cell events or exclusion of neoplastic PCs (please see
20 events is a conservative value for the smallest (homoge- PC gating above).
neous) population that can be detected in a given flow  Variation (e.g.: increase) in minor antigen profiles on
cytometric list mode data file by experienced operators normal PC subsets (e.g.: CD192, CD452, and
(26,27). Therefore, the maximum number of events that CD561); please note that there are no specific

Cytometry Part B: Clinical Cytometry


38 ARROZ ET AL.

individual tumor antigens, and one marker alone is 3. van Dongen JJM, Lhermitte L, B€ ottcher S, Almeida J, van der Velden
V, Flores-Montero J, Rawstron A, Asnafi V, L ecrevisse Q, Lucio P,
typically not enough to define neoplastic PCs. et al. EuroFlow antibody panels for standardized n-dimensional flow
 Presence of minor PC subpopulations that only cytometric immunophenotyping of normal, reactive and malignant
become apparent with the acquisition of >106 BM leukocytes. Leukemia 2012;26:1908–1975
cells. 4. Cannizzo E, Bellio E, Sohani AR, Hasserjian RP, Ferry JA, Dorn ME,
Sadowski C, Bucci JJ, Carulli G, Preffer F. Multiparameter immuno-
 Phenotypic heterogeneity of neoplastic PCs at diagno- phenotyping by flow cytometry in multiple myeloma; the diagnostic
sis should be taken into account to differentiate anti- utility of defining ranges of normal antigenic expression in compari-
son to histology. Cytometry B Clin Cytom 2010;78:231–238.
gen shifts within neoplastic PCs (e.g.: loss of CD56),
5. Liu D, Lin P, Hu Y, Zhou Y, Tang G, Powers L, Medeiros LJ,
from phenotypic selection of chemo-resistant subclones Jorgensen JL, Wang SA. Immunophenotypic heterogeneity of normal
(antigen restriction rather than shift). plasma cells: Comparison with minimal residual plasma cell
 Neoplastic PCs do not have predictable FSC/SSC and myeloma. J Clin Pathol 2012;65:823–829
6. Tembhare PR, Yuan CM, Venzon D, Braylan R, Korde N, Manasanch
CD45/SSC profiles and they often show substantial E, Zuchlinsky D, Calvo K, Kurlander R, Bhutani M, et al. Flow cyto-
autofluorescence. metric differentiation of abnormal and normal plasma cells in the
bone marrow in patients with multiple myeloma and its precursor
diseases. Leuk Res 2014;38:371–376
SUMMARY OF MINIMUM RECOMMENDATIONS
7. Cao W, Goolsby CL, Nelson BP, Singhal S, Mehta J, Peterson LC.
Instability of immunophenotype in plasma cell myeloma. Am J Clin
Pathol 2008;129: 926–933
 Minimum requirements for flow-MRD monitoring in 8. Rawstron AC, Child JA, de Tute RM, Davies FE, Gregory WM, Bell
MM include the usage of at least six-color antibody SE, Szubert AJ, Navarro-Coy N, Drayson MT, Feyler S, et al. Minimal
residual disease assessed by multiparameter flow cytometry in mul-
combinations in a single tube, most preferably eight, a tiple myeloma: Impact on Outcome in the Medical Research Coun-
gating strategy based on simultaneous assessment of cil Myeloma IX Study. J Clin Oncol 2013;31:2540–2547.
CD38, CD138, and CD45 expression to maximize PC 9. Paıno T, Paiva B, Sayagues J, Mota I, Carvalheiro T, Corchete L,
Aires-Mejia I, Perez JJ, Sanchez ML, Barcena P, et al. Phenotypic
inclusion whilst minimizing contamination by other identification of subclones in multiple myeloma with different che-
cells along with the use of FSC and SSC to exclude moresistant, cytogenetic and clonogenic potential. Leukemia (in
cell doublets and debris, and at least one combination press). doi:10.1038/leu.2014.321
10. Wood BL, Arroz M, Barnett D, DiGiuseppe J, Greig B, Kussick SJ,
that simultaneously includes CD38/CD45/CD19/ Oldaker T, Shenkin M, Stone E, Wallace P. 2006 Bethesda Interna-
CD56. tional Consensus Recommendations on the Immunophenotypic
 Interpretation of antigen expression profiles should Analysis of Hematolymphoid Neoplasia by Flow Cytometry: Optimal
Reagents and Reporting for the Flow Cytometric Diagnosis of
be performed with caution as many of the reported Hematopoietic Neoplasia. Cytometry B Clin Cytom 2007;72B:S14–
aberrancies based on 1-2D marker analysis are also S22
present in specific (e.g., minor) subsets of normal 11. Wood B. 9-Color and 10-Color Flow Cytometry in the Clinical Labo-
ratory. Arch Pathol Lab Med 2006;130:680–686
PCs.
12. Costa ES, Pedreira CE, Barrena S, Lecrevisse Q, Flores J, Quijano S,
 The capacity to perform additional analysis of samples Almeida J, del Carmen Garcıa-Macias M, Bottcher S, Van Dongen JJ,
that require further evaluations by either acquisition et al. Automated pattern-guided principal component analysis VS
of more events and/or characterization of further anti- expert-based immunophenotypic classification of B-cell chronic lym-
phoproliferative disorders: A step forward in the stardardization of
gens namely CyIgj/CyIgk on the same sample while it Immunophenotyping. Leukemia 2010;24:1927–1933
still remains relatively fresh, is recommended in cen- 13. Liu D, Lin P,HuY, Zhou Y,Tang G, Powers L, Medeiros J, Jorgensen J,
ters that perform MRD assessment. Wang S. Immunophenotypic heterogeneity of normal plasma cells:
Comparison with minimal residual plasma cell myeloma. J Clin
 The role of cytoplasmic immunoglobulin light chain Pathol 2012;65:823–829.
j/k expression for high-sensitive MRD detection 14. Kozbor D, Moretta A, Messner H, Moretta L, Croce C. Tp44 mol-
(1025) is currently being evaluated. ecules involved in antigen-independent T cell activation are
expressed on human plasma cells. J Immunol 1987;138:4128–
 The report should include the MRD level plus the 4132.
LOD and LLOQ of the assay, the proportion and num- 15. Aghaeepour N, Finak G, FlowCAP Consortium; DREAM Consortium,
ber of neoplastic and normal PCs (if present) within Hoos H, Mosmann TR, Brinkman R, Gottardo R, Scheuermann RH.
the BM PC compartment, the immunophenotypic Critical assessment of automated flow Cytometry data techniques.
Nat Methods 2013;10:228–238
expression profile of neoplastic PCs, as well as cell 16. Pedreira CE, Costa ES, Lecrevisse Q, van Dongen JJM, Orfao A.
viability and the quality of sample (e.g.: percentages Overview of clinical flow cytometry data analysis: Recente advances
of B-cell precursors, nucleated red cells, and/or mast and future challenges. Trends Biotechnol 2013;31:415–425
cells to reflect potential hemodilution based on the 17. Pedreira CE, Costa ES, Barrena S, Lecrevisse Q, Almeida J, van
Dongen JJ, Orfao A. Generation of flow cytometry data files with a
absence of such BM-specific cell populations). potentially infinite number of dimensions. Cytometry A 2008;73:
834–846
LITERATURE CITED 18. Paiva B, Martinez-Lopez J, Vidriales MB, Mateos MV, Montalban MA,
Fernandez-Redondo E, Alonso L, Oriol A, Teruel AI, de Paz R, et al.
1. Tanqri S, Vall H, Kaplan D, Hoffman B, Purvis N, Porwit A, Comparison of immunofixation, serum free light chain, and immu-
Hunsberger B, Shankey T. Validation of cell-based fluorescence nophenotyping for response evaluation and prognostication in mul-
assays: Practice guidelines from the ICSH and ICCS – Part III – ana- tiple myeloma. J Clin Oncol 2011;29:1627–1633
lytical issues. Cytometry B Clin Cytom 2013;84:291–308. 19. Paiva B, Gutierrez NC, Rosinol L, Vıdriales MB, Montalban MA ,
2. Rawstron AC, Orfao A, Beksac M, Berdickova L, Brooimans R, Martınez-Lopez J, Mateos MV, Cibeira MT, Cord on L, Oriol A, et al.
Bumbea H, Dalva K, Fuhler G, Gratama J, Hose D, et al. Report of High-risk cytogenetics and persistent minimal residual disease by
the European Myeloma Network on multiparametric flow Cytome- multiparameter flow cytometry predict unsustained complete
try in multiple myeloma and related disorders. Haematologica 2008; response after autologous stem cell transplantation in multiple
93:431–438. myeloma. Blood 2012;119:687–691

Cytometry Part B: Clinical Cytometry


GUIDELINES ON MYELOMA MRD ANALYSIS AND REPORTING 39

20. Puig N, Sarasquete ME, Balanzategui A, Martınez J, Paiva B, Garcıa 26. Hedley BD, Keeney M. Technical issues: Flow cytometry and rare
H, Fumero S, Jimenez C, Alcoceba M, Chill on MC, et al. Critical event analysis. Int J Lab Hematol 2013;35:344–350.
evaluation of ASO RQ-PCR for minimal residual disease evaluation 27. Subira D,Casta~
non S, Aceituno E, Hernandez J, Jimenez-Gar
ofan C,
in multiple myeloma. A comparative analysis with flow cytometry. Jimenez A, Jimenez AM, Roma A, Orfao A. Flow cytometric analysis
Leukemia 2014;28:391–397. of cerebrospinal fluid samples and its usefulness in routine clinical
21. Paiva B, Vidriales MB, Cervero J, Mateo G, Perez JJ, Montalban MA, practice. Am J Clin Pathol 2002;117:952–958.
Sureda A, Montejano L, Gutierrez NC, Garcıa de Coca A, et al. Multi- 28. Rawstron A, B€ ottcher S, Letestu R, Villamor N, Fazi C, Kartsios
parameter flow cytometric remission is the most relevant prognos- H, de Tute RM, Shingles J, Ritgen M, Moreno C, et al. Improving
tic factor for multiple myeloma patients who undergo autologous efficiency and sensitivity: European Research Initiative in CLL
stem cell transplantation Blood 2008;112:4017–4023 (ERIC) update on the international harmonised approach for flow
22. Domingo E, Moreno C, Sanchez-Ibarrola A, Panizo E, Paramo JA, cytometric residual disease monitoring in CLL. Leukemia 2013;27:
Merino J. Enhanced sensitivity of flow cytometry for routine 142–149.
assessment of minimal residual disease. Haematologica 2010;95: 29. Nieto WG, Almeida J, Romero A, Teod osio C, L opez A,
691–692 Henriques AF, Sanchez ML, Jara-Acevedo M, Basilio A, Gonzalez
23. College of American Pathologists (CAP). Laboratory Accreditation M, et al. Increased frequency (12%) of circulating chronic lym-
Manual 2012 Edition. Illinois: College of American Pathologists; 2012. phocitic Leukemia-like B-cell clones in healthy subjects using a
24. Clinical and Laboratory Standards Institute (CLSI). Evaluation of highly sensitive multicolor flow cytometry approach. Blood 2009;
Detection Capability for Clinical Laboratory Measurement Proce- 114:33–37
dures; Approved Guideline, 2nd ed. CLSI document EP17-A2 (ISBN 30. Paiva B, Vidriales MB, Mateo G, Perez JJ, Montalban MA, Sureda A,
1-56238-795-2). Pennsylvania: Clinical and Laboratory Standards Montejano L, Guti errez NC, Garcıa de Coca A, de las Heras N, et al.
Institute; 2012. The persistence of immunophenotypically normal residual bone
25. Jennings L, Van Deerlin VM, Gulley ML. Recommended practices marrow plasma cells at diagnosis identifies a good prognostic sub-
and principles for validating clinical molecular pathology tests. Arch group of symptomatic multiple myeloma patients Blood 2009;114:
Pathol Lab Med 2009;133:743–755 4369–4372

Cytometry Part B: Clinical Cytometry

Potrebbero piacerti anche